61 1

Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R13–R24 Endocrinology endocrine cells REVIEW Epigenetic regulation in the tumorigenesis of MEN1-associated endocrine cell types

Sucharitha Iyer and Sunita K Agarwal

Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA

Correspondence should be addressed to S K Agarwal: [email protected]

Abstract

Epigenetic regulation is emerging as a key feature in the molecular characteristics of Key Words various human diseases. Epigenetic aberrations can occur from mutations in ff neuroendocrine associated with epigenetic regulation, improper deposition, removal or reading of ff pancreas histone modifications, DNA methylation/demethylation and impaired non-coding ff parathyroid RNA interactions in chromatin. Menin, the product of the causative for ff pituitary the multiple endocrine neoplasia type 1 (MEN1) syndrome, interacts with chromatin- associated protein complexes and also regulates some non-coding RNAs, thus participating in epigenetic control mechanisms. Germline inactivating mutations in the MEN1 gene that encodes menin predispose patients to develop endocrine tumors of the parathyroids, anterior pituitary and the duodenopancreatic neuroendocrine tissues. Therefore, functional loss of menin in the various MEN1-associated endocrine cell types can result in epigenetic changes that promote tumorigenesis. Because epigenetic changes are reversible, they can be targeted to develop therapeutics for restoring the tumor epigenome to the normal state. Irrespective of whether epigenetic alterations are the cause or consequence of the tumorigenesis process, targeting the endocrine tumor- associated epigenome offers opportunities for exploring therapeutic options. This review presents epigenetic control mechanisms relevant to the interactions and targets of menin, and the contribution of epigenetics in the tumorigenesis of endocrine cell types Journal of Molecular from menin loss. Endocrinology (2018) 61, R13–R24

Introduction

Epigenetic changes observed in various human diseases regulation in tumor etiology is highlighted by the have been shown to play important roles as disease discovery of mutations in genes encoding factors that drivers, biomarkers and therapeutic targets. Epigenetic participate in epigenetic regulatory mechanisms in changes can occur due to mutations in factors that various cancers (Koschmann et al. 2017). Menin, the control epigenetic modifications or in response to protein encoded by the gene responsible for causing the metabolic or environmental signals, which lead to multiple endocrine neoplasia type 1 (MEN1) syndrome, improper deposition, removal or reading of histone participates in epigenetic regulation (Shilatifard 2012, van modifications, DNA methylation/demethylation and Nuland et al. 2013). Patients with the MEN1 syndrome impaired non-coding RNA interactions in chromatin carry germline heterozygous inactivating mutations (Allis et al. 2015). The evidence for the role of epigenetic in the MEN1 gene located at 11q13, that

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access

-18-0050 Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R14 Endocrinology endocrine cells encodes menin, which predisposes to the development of could potentially promote tumorigenesis. Furthermore, tumors in multiple endocrine organs (Chandrasekharappa evidence from genome and exome sequencing efforts et al. 1997, Lemmens et al. 1997). The main MEN1- of sporadic non-functioning and functioning PNETs, associated endocrine cell types that form tumors are parathyroid adenoma and lung neuroendocrine tumors located in the parathyroids, anterior pituitary and the have shown additional somatic mutations in other genes duodenopancreatic neuroendocrine tissues (Thakker associated with epigenetic control mechanisms that et al. 2012). Other MEN1-associated endocrine tumors predict epigenetic changes to promote tumorigenesis in include foregut carcinoids that arise from neuroendocrine these endocrine cell types (Jiao et al. 2011, Cromer et al. cells of the lung, thymus or gastrointestinal (GI) tract. 2012, Chou et al. 2016, Romano et al. 2017, Scarpa et al. The tumors are classified as functioning when they 2017, Simbolo et al. 2017, Wang et al. 2017). hypersecrete the hormones characteristic of the endocrine Menin is a 67 kDa ubiquitously expressed cell types affected, or non-functioning when they do predominantly nuclear protein, with no intrinsic not hypersecrete any hormones. Parathyroid tumors enzymatic activity. As stated earlier, menin is a tumor are functioning causing primary hyperparathyroidism suppressor in the context of MEN1-associated endocrine due to excessive parathyroid hormone (PTH) secretion. cell types; however, menin has been shown to act as a pro- Pituitary and duodenopancreatic neuroendocrine tumors oncogenic co-factor in mixed lineage leukemia (Yokoyama in MEN1 patients are observed to be either functioning et al. 2005, Muntean & Hess 2012). The crystal structure (prolactinoma, 20%; somatotropinoma, 10%; gastrinoma, of menin shows a deep pocket that can act as a binding 40%; insulinoma, 10%) or non-functioning (pituitary, site for interacting ( No. 3RE2 5%; PNETs, 20–55%) (Schussheim et al. 2001, Thakker and 3U84) (Murai et al. 2011, Huang et al. 2012). Evidence et al. 2012, Marini et al. 2017). In the context of these from protein–protein interaction studies have shown endocrine tumors, MEN1 acts as a classic tumor suppressor that menin is involved in epigenetic regulation and gene because human MEN1-associated tumors show loss gene transcription through interaction with proteins in of heterozygosity (LOH) at 11q13 resulting in biallelic chromatin-associated protein complexes and transcription loss/inactivation of MEN1 (Chandrasekharappa et al. factors, affecting the expression of target genes such as 1997, Lemmens et al. 1997). Similarly, in genetically genes that control cell proliferation (Karnik et al. 2005, Wu engineered mouse models, germline targeted deletion of & Hua 2011). Interacting partners of menin also include both copies of the Men1 gene causes embryonic lethality, proteins that participate in cell adhesion, cell division, while germline targeted deletion of one copy of the cell motility, cell signaling, cytoskeletal structure, DNA Men1 gene generates live mice that later in life develop repair in response to DNA damage and studies conducted endocrine tumors similar to those seen in the human in Drosophila melanogaster have shown a role of menin MEN1 syndrome with LOH at the Men1 (Agarwal in the maintenance of genomic stability (Busygina et al. 2014, Agarwal 2017b). 2004, Hendy et al. 2009, Agarwal 2017a, Feng et al. 2017a). Interestingly, somatic MEN1 mutations are also Investigations to elucidate the biological function of detected in 30–40% of sporadic tumors of MEN1- menin through protein–protein interaction studies have associated endocrine cell types – parathyroid adenomas, significantly advanced our knowledge about the potential gastrinomas, pancreatic neuroendocrine tumors (PNETs) roles of menin in various cellular pathways, but they have and lung neuroendocrine tumors (Zhuang et al. 1997, been mainly conducted using cell lines (HEK293, HeLa, Debelenko et al. 1997, Jiao et al. 2011, Cromer et al. 2012, HepG2, K562 and mouse embryo fibroblasts (MEFs)) that Newey et al. 2012, Scarpa et al. 2017, Simbolo et al. 2017). are unrelated to MEN1-associated endocrine cell types. Among sporadic PNETs, MEN1 mutations are detected in Follow-up functional characterization of some interactions the non-functioning PNETs, but rarely in the functioning of menin have been performed in endocrine/endocrine- PNETs (insulinomas) (Jiao et al. 2011, Cao et al. 2013, related cell lines (βlox5, BON1, INS-1, MIN6, NIH-H720, Lichtenauer et al. 2015, Chou et al. 2016, Scarpa et al. NIH-727, and QGP1) albeit not in endocrine cell lines 2017, Wang et al. 2017). Also, MEN1 mutations are rarely with MEN1 mutations or that lack menin because no such detected in sporadic pituitary tumors (Poncin et al. 1999, cell lines are currently available. The focus of this review Newey et al. 2013, Bi et al. 2017, Marques & Korbonits will be on studies conducted using endocrine/endocrine- 2017). Because menin participates in epigenetic regulation, related cell lines or menin-null endocrine tumors from functional loss of menin in the various MEN1-associated mouse models. This review excludes the discussion of endocrine cell types would lead to epigenetic changes that menin’s interaction with transcription factors because

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R15 Endocrinology endocrine cells they were recently well covered elsewhere (Agarwal (Zoghbi & Beaudet 2016). Technological advances in 2017a, Dreijerink et al. 2017, Feng et al. 2017a). In this chromatin immunoprecipitation and next-generation review, we present a survey of the published literature to sequencing have allowed comprehensive characterization describe epigenetic control mechanisms that are relevant of genome-wide epigenetic marks to reveal the status of to the interactions and targets of menin, the contribution the epigenome in normal and pathological conditions that of epigenetics in the tumorigenesis of endocrine cell types can help to identify epigenomic changes that are disease from menin loss and experimental therapeutic strategies drivers or to serve as biomarkers and therapeutic targets. to potentially correct the tumor-specific epigenetic Below we describe the various epigenetic mechanisms changes. and chromatin modifications that are relevant to MEN1- associated endocrine cell types (Fig. 1). Epigenetic mechanisms

Eukaryotic DNA with core histone proteins (H2A, H2B, H3 and H4) is packaged into nucleosomes to form chromatin fibers (Olins & Olins 2003). This packaging regulates access to DNA for various biological processes such as DNA replication, DNA repair and transcription into RNA. Modifications to chromatin are essential to permit or restrict access, for example, to the transcriptional machinery including transcription factors for gene expression (Flavahan et al. 2017). Chromatin modifiers and remodelers constitute ‘epigenetic factors’ that function in multi-protein complexes that may also include non- coding RNAs, to modify the DNA or histones contained in nucleosomes (Allis & Jenuwein 2016). The multi- protein complexes contain enzymes and proteins referred to as the ‘writers’, ‘readers’ and ‘erasers’ of chromatin modifications Allis( & Jenuwein 2016). Modifications to DNA include methylation, hydroxymethylation and further oxidation. Histones can undergo a variety of post- translational modifications (PTMs) such as methylation, acetylation, phosphorylation and ubiquitination to generate an active, repressive or poised state to control gene expression; and the array of PTMs that govern a specific chromatin state is referred to as the ‘histone code’ (Huang et al. 2014). Also, chromatin interactions can occur over short and long distances thereby generating conditions that enhance gene transcription or insulate the regions of gene transcription (Rowley & Corces 2016). ‘Epigenetic changes’ through alterations in DNA and histone modifications, chromatin remodeling and alterations in non-coding RNA interactions, create a Figure 1 Overview of menin-dependent epigenetic regulation in MEN1-associated positive or negative environment affecting the outcome endocrine cell types. Epigenetic mechanisms and the observed or of gene transcription and chromatin structure (Allis et al. predicted consequence of menin loss are shown that were investigated in 2015). The ‘epigenetic marks’ on DNA and histones that various studies of human or mouse endocrine tumors or studies conducted in endocrine/endocrine-related cell lines. Green or red are generated as a result of the enzymatic activity of various indicates the normal effect of the epigenetic event resulting in the chromatin-modifying enzymes can survive successive activation or repression of gene expression, respectively. Up or down cell divisions, and they are reversible, which presents arrow indicates increase or decrease, respectively, of the epigenetic event in a cell line or tumor with menin deficiency. ALT (alternative lengthening an opportunity for correction to the normal state, for of telomeres) is shown in blue (and not in red or green) because it is an example, when such changes promote disease processes event associated with chromatin structure at telomeres.

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R16 Endocrinology endocrine cells

Histone methylation and demethylation Interestingly, both KMT2A and MEN1 co-localize at human chromosome 11q and mouse chromosome 19. LOH at Methylation of histones at lysine or arginine residues this locus in MEN1 mutation-positive human PNETs and in the chain of amino acids that protrude from the mouse PNETs (insulinomas) in Men1+/− mice contributes to nucleosome, referred to as the ‘histone tail’, is achieved the tumorigenesis process. Targeted conditional deletion by histone methyltransferases (KMTs) that exist in of both Kmt2a and Men1 in mice (Kmt2af/f;Men1f/f;RIP-Cre) multi-subunit protein complexes (Cheng 2014). KMTs led to the acceleration of pancreatic islet tumor formation methylate histone H3 on lysine residues at positions 4, 9, and also shortened the average life span of these mice (Lin 27, 36 and 79 in the in mono-, di- or tri-methylated form et al. 2016). This study showed that MLL1 can cooperate depending on whether one, two or three methyl groups with menin to prevent tumor formation. In another study, are added, respectively (Chi et al. 2010). In general, the targeted conditional deletion in mice of Men1 and Rbp2, histone H3 lysine-4 trimethyl mark (H3K4me3) specifies which encodes a H3K4me3 demethylase (Rbp2f/f;Men1f/ a state of transcriptional activation, which is located at f;RIP-Cre) decreased the rate of tumor formation and the transcriptional start site (TSS) of actively transcribed prolonged survival (Lin et al. 2011). These data showed genes. On the other hand, H3K9me3 and H3K27me3 that the histone demethylase activity of RBP2 is linked to are associated with transcriptional silencing. When both islet tumorigenesis downstream of menin loss. H3K4me3 and H3K27me3 occur together, the gene is said To investigate the contribution of menin-MLL1- to be bivalently poised – a state maintained, for example, mediated epigenetic regulation in tumor formation, by embryonic stem cells during lineage differentiation and genome-wide distribution of the gene activation mark development, to enable rapid switching between active H3K4me3 and its counterpart recessive mark H3K27me3 and repressive gene transcription states (Voigt et al. 2013). was examined in menin-deficient pancreatic islets of Eraser KDMs such as lysine-specific demethylase 1 (LSD1/ 2-month-old Men1f/f;RIP-Cre and control wild-type KDM1A), lysine-specific demethylase 2 (LSD2/KDM1B) mice (Lin et al. 2015). This study found that within and Jumonji AT-rich interacting domain 1A (JARID1A/ the same set of genes, loss of H3K4me3 correlated with KDM5A/RBP2), remove mono- , di- or tri-methylation of increased H3K27me3 in menin-deficient islets, and the H3K4 or H3K9 (Cheng 2014). Depending on the specific expression of such genes was downregulated. The most amino acid residue methylated or demethylated, and the significantly downregulated gene in menin-deficient level of methylation, the process of histone methylation islets was Igfbp2, which also displayed loss of H3K4me3 and demethylation generates active or repressive together with gain of H3K27me3. Interestingly, the chromatin states. decline in Igfbp2 expression in menin-deficient islets could be reversed with simultaneous deletion of Rbp2 (Lin et al. 2015). This observation further confirms that H3K4me3 and H3K27me3 the histone demethylase activity of RBP2 is linked to islet There are at least six different protein complexes tumorigenesis downstream of menin loss. that contain KMTs for catalyzing the gene-activating Genome-wide occupancy patterns of menin, MLL1 histone modification H3K4me3 Dreijerink( et al. 2017). and another component of the MLL-protein-complex Menin has been shown to interact with KMTs, MLL1 (RBBP5) in human pancreatic islet cells, combined with (KMT2A) and MLL2 (KMT2B), in two of those multi- gene expression analysis in menin-null islets from Men1f/ subunit protein complexes, and studies using chromatin f;RIP-Cre mice showed upregulation of the pancreatic immunoprecipitation have shown that menin is present islet β-cell differentiation factor HLXB9 (Scacheri et al. at the TSS with H3K4me3 (Hughes et al. 2004, Yokoyama 2006). Candidate gene analysis using menin-null islets et al. 2004, Scacheri et al. 2006). Several lines of evidence from Men1f/f;RIP-Cre mice showed downregulation of have established the importance of this interaction of cell cycle inhibitory genes in menin-null islet tumors menin involving H3K4me3 and the target genes affected compared to normal islets, mainly the cyclin-dependent using MEN1-associated endocrine cell types, mainly kinase inhibitor (CDKI) genes CDKN2C/p18 and pancreatic islets. CDKN1B/p27, with concomitant loss of H3K4me3 in Mice with targeted deletion of Men1 in the germline their promoter regions (Karnik et al. 2005). In another (Men1+/−) or specifically in the pancreatic isletβ -cells study, a long non-coding RNA, MEG3 was identified as an (Men1f/f;RIP-Cre) develop insulin-secreting pancreatic epigenetic target of menin (Agarwal & Jothi 2012). This islet tumors (insulinomas) (Agarwal 2014, 2017b). study used genome-wide assessment of H3K4me3 and

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R17 Endocrinology endocrine cells gene expression from wild-type mouse embryonic stem and symmetric dimethylation of arginines. In mammalian cells (mESCs) and menin-null mESCs. Loss of H3K4me3 cells, PRMT5 is the predominant type II arginine was observed in menin-null mESCs at the Meg3 gene, methyltransferase, which acts as a transcriptional together with downregulation of the Meg3 transcript. repressor specifying H3R2me2 and H4R3me2 (Stopa et al. Further studies showed decreased MEG3 levels in mouse 2015). Menin has been shown to interact with PRMT5, and human islet tumors (Modali et al. 2015). Also, loss coinciding with a repressive chromatin mark, symmetric of MEG3 expression was reported in sporadic non- dimethylation of H4R3 (H4R3me2s), at the promoter of the functioning pituitary tumors (Zhao et al. 2005). When growth arrest-specific 1 (Gas1) gene (Gurung et al. 2013). mESCs were in vitro differentiated into pancreatic islet like GAS1 has been shown to direct the binding of the sonic endocrine cells (PILECs), reduced H3K4me3 and reduced hedgehog (SHH) ligand to its receptor patched 1 (PTCH1) gene expression was observed of the Hox genes in PILECs to activate the pro-proliferative and oncogenic Hedgehog derived from menin-null mESCs (Agarwal & Jothi 2012). signaling pathway (Martinelli & Fan 2007). Thus, menin HOX genes have been identified as the direct target of the loss would coincide with increased GAS1 protein and MLL1-complex in many different cell types, particularly oncogenic Hedgehog signaling. Further characterization in hematopoetic cells where the menin-MLL association of menin/PRMT5 showed that pharmacologic inhibition partners as an oncogenic complex in MLL-fusion leukemia of Hedgehog signaling with the drug GDC-0449 reduced (Muntean & Hess 2012). It is important to note that the cell proliferation in insulinoma tumors of Men1f/f;RIP- PILECs were heterogeneous in terms of the cell population Cre mice (Gurung et al. 2013). These data show that as assessed by the expression of pancreatic marker genes, inhibition of Hedgehog signaling in PNETs could target and not entirely consisted of pancreatic endocrine cells the downstream effects from loss of menin/PRMT5 (Agarwal & Jothi 2012). Whether HOX genes play a role interaction, revealing a potential target for treating MEN1 in MEN1-associated endocrine tumorigenesis remains to tumors. be determined. Histone acetylation and deacetylation H3K9me3 Histone acetyl transferases (HATs) catalyze acetylation Another histone lysine modification regulated by menin at lysine residues in histone tails. There are five well- is the repressive H3K9me3, through interaction with a characterized HAT families that can acetylate specific lysine methyltransferase, suppressor of variegation 3–9 lysine residues of mammalian histones – p300/CBP, Gcn5- homolog 1 (SUV39H1) (Yang et al. 2013). In INS-1 rat related N-acetyltransferase (GNAT) (GCN5/PCAF), MYST insulinoma cells (Merglen et al. 2004), enhanced H3K9me3 (for MOZ, Ybf2/Sas3, Sas2 and Tip60)-related HATs, the was detected at the promoter region of the membrane general transcription factor HATs, which include the TFIID metalloproteinase (Mme) gene with combined presence of subunit TAF250 and nuclear/steroid hormone receptor menin, SUV39H1 and the chromatin-remodeling protein coactivators (SRCs) (Marmorstein & Zhou 2014). The DAXX (Feng et al. 2017b). The DAXX gene is frequently acetylation marks on histones are biologically read by the mutated in sporadic non-functioning PNETs (Jiao et al. bromodomain (BRD) contained in the bromodomain and 2011, Chou et al. 2016, Scarpa et al. 2017). Therefore, extraterminal (BET) proteins comprising members BRD2, this study helps to integrate the potential involvement of BRD3, BRD4 and BRDT. Histone acetylation generally multiple epigenetic regulators to prevent tumorigenesis. favors active transcription that is reversed by the activity of histone deacetylases (HDACs) (Marmorstein & Zhou 2014). There are 18 HDACs that belong to four different H4R3me2s subtypes classified on sequence similarities, shared PRMTs methylate arginine residues in histone H3 at catalytic mechanisms and active sites (Seto & Yoshida positions 2, 8, 17 and 26; and histones H4 and H2A at 2014). Small-molecule inhibitors (CPI-203, JQ1 and PFI- positions 3 and 11, respectively (Yang & Bedford 2013). 1) of bromodomain interactions with acetylated histone PRMTs are classified into two families. The type I family tails have been developed as potential therapeutics (Perez- comprises the members 1, 2, 3, 4, 6 and 8, which catalyze Salvia & Esteller 2017). JQ1 binds to the acetyl-lysine mono-methylation and asymmetric dimethylation recognition pocket of bromodomains 1 and 2 on BRD4, of arginines (Yang & Bedford 2013). The type II family which displaces BRD4 from chromatin, thus preventing consisting of PRMTs 5, 7 and 9 mediate monomethylation the ‘reading’ of the acetyl mark and blocking active

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R18 Endocrinology endocrine cells transcription. CPI-203 is a primary amide analog of JQ1, The evidence for the involvement of histone and PFI-1 inhibits BRD2 and BRD4 (Perez-Salvia & Esteller remodeling in MEN1-associated endocrine cells comes 2017). from sporadic non-functioning PNETs that show somatic There is no direct evidence in MEN1-associated mutations in death-domain-associated protein (DAXX) endocrine cells for the interaction of menin in protein and its interacting partner, alpha thalassemia/mental complexes that are involved in specific histone acetylation retardation syndrome X-linked (ATRX) (Jiao et al. 2011, or deacetylation. However, there is indirect evidence for Chou et al. 2016, Scarpa et al. 2017). ATRX with its the involvement of histone acetylation/deacetylation in intrinsic ATPase activity, in collaboration with its partner endocrine cells that comes from the demonstration of DAXX, a highly specific histone chaperone, deposit the growth inhibitory effects of HDAC inhibitors (HDCAi) histone variant H3.3 in chromatin at telomeric and (trichostatin-A (TSA), thailandepsin‐A (TDP‐A), sodium pericentromeric regions (Amorim et al. 2016). Telomeres butyrate (NaB), valproic acid (VPA) and MS-275), and BET protect chromosome ends to prevent the loss of DNA, protein bromodomain inhibitors (BETi) (CPI201, PFI-1 and in neoplastic cells, the maintenance of telomeres is and JQ1) in neuroendocrine tumor cell lines BON1, NCI- required for immortality. Lengthening of telomeres in H720, NCI-H727 and QGP1 (Baradari et al. 2006, Adler tumor cells can occur from excessive production of the et al. 2009, Wong et al. 2014, Arvidsson et al. 2016, Lines telomerase enzyme or through alternative lengthening et al. 2017). BON1 is a human cell line derived from a of telomeres (ALT), a process similar to homologous peripancreatic lymph node metastasis of a pancreatic recombination (Amorim et al. 2016). ATRX/DAXX neuroendocrine carcinoid tumor (Townsend et al. 1993). mutations or loss of their expression in sporadic PNETs NCI-H720 and NCI-H727 are derived from human atypical correlate with increased telomere length that can and typical bronchial carcinoid tumors, respectively potentially support tumor progression (Kim et al. 2017, (ATCC). QGP-1 is a carcinoembryonic antigen-producing Marinoni et al. 2017, Scarpa et al. 2017, Singhi et al. 2017, cell line from a human pancreatic islet carcinoma (Kaku VandenBussche et al. 2017). Some level of ALT was also et al. 1980). Anti-tumor activity of the HDACi VPA and detected in MEN1 mutation-positive sporadic PNETs BETi CPI203 was also shown in a BON1 xenograft model (Scarpa et al. 2017). Whether ALT plays a role in the (Greenblatt et al. 2007, Wong et al. 2014). Also, in vivo anti- endocrine tumors of patients with the MEN1 syndrome tumor activity of JQ1 has been demonstrated in primary remains to be determined. tumors (insulinomas) of the Men1f/f;RIP-Cre mouse model (Lines et al. 2017). These studies highlight the potential of DNA methylation and demethylation targeting the epigenetic histone acetyl marks in treating MEN1-associated neuroendocrine tumors. DNA methylation is an epigenetic modification that occurs predominantly at CpG sites where a cytosine nucleotide is followed by a guanine nucleotide in which Chromatin remodeling the cytosine is methylated to 5-methylcytosine (5-mC) (Li Histones and nucleosomes are subjected to remodeling & Zhang 2014). Methylation at CpG islands (GC-rich DNA by protein complexes that contain ATP-dependent regions) is often located in promoter regions of genes. chromatin-remodeling activity. Also, eukaryotic cells Aberrant hypo- or hyper-methylation of these promoter express histone variants, in addition to the ‘canonical’ region CpG islands can result in gene overexpression core histones, providing another layer of epigenetic or gene silencing, respectively. A family of DNA methyl regulation. Chromatin-remodeling activity facilitates transferases (DNMTs), designated DNMT1, DNMT3A and transcriptional initiation and elongation for the passage DNMT3B, mediates CpG methylation. While DNMT3A of RNA polymerase through nucleosomal arrays (Clapier and DNMT3B establish de novo DNA methylation, & Cairns 2009). This is achieved by displacing histone DNMT1 serves to propagate and stably maintain the DNA dimers H2A/H2B or H3/H4 leading to the substitution of methylation mark (Li & Zhang 2014). DNA methylation the core histones in the dimers with histone variants such can be reversed by the ten-eleven translocation (TET) as histone H2A.Z or H3.3 (Clapier & Cairns 2009, Szenker family of methylcytosine hydroxylases that convert et al. 2011). In addition to controlling gene transcription, 5mC to 5-hydroxymethylcytosine (5-hmC) (Li & Zhang histone variants and their PTMs also contribute to 2014). Also, drugs such as 5-azacitidine (5AC) and 5-aza- nucleosome and chromatin structure and function 2′-deoxycytidine (DAC) can block DNA methylation by (Szenker et al. 2011). directly inhibiting DNMTs (Sato et al. 2017).

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R19 Endocrinology endocrine cells

The histone modification H3K4me3 has been the potential benefit of DNA methylation inhibitors in shown to protect CpG islands from DNA methylation treating PNETs, pituitary tumors and parathyroid tumors. resulting in an antagonism between H3K4me3 and DNA methylation to regulate gene transcription (Cedar & Non-coding RNA interactions Bergman 2009). Also, DNA methylation in transcription factor recognition sequences can alter transcription factor Non-coding RNAs (ncRNAs), as the name indicates, lack binding (Barlow & Bartolomei 2014). A study conducted open reading frames (ORFs) and do not encode proteins. in MIN6 mouse insulinomas cells (Miyazaki et al. 1990) Short ncRNAs such as microRNA (miRNA) are processed showed that overexpression of menin increased the from longer primary transcripts into small single-stranded expression of the lncRNA Meg3 (Modali et al. 2015). This molecules (20–24 nucleotides). They perform RNAi- effect coincided with H3K4me3 at the Meg3 promoter, and mediated (RNA interference-mediated) gene silencing hypomethylation of the central CpG of a cAMP response by complementary base-pair interactions targeting DNA element (CRE) that binds the transcription factor CREB; in chromatin for RNA-induced transcriptional silencing the DNA hypomethylation prevented CREB binding (RITS), or messenger RNAs (mRNAs) post-transcriptionally (Zhao et al. 2006, Modali et al. 2015). Whether similar for degradation or by blocking translation (Cech & Steitz instances of reciprocal H3K4me3 and DNA methylation 2014). Long non-coding RNAs (lncRNAs) are transcripts together with altered transcription factor binding are of 200 bp or more in length that can interact with DNA present in MEN1-associated endocrine cells remains to be and proteins whereby they also participate in epigenetic determined. regulation (Kopp & Mendell 2018). Some lncRNAs have Several studies have shown indirect evidence for the been shown to reside in chromatin-modifying and participation of DNA methylation in MEN1-assocated chromatin-remodeling protein complexes that silence endocrine cell types. DNA hypermethylation has been gene expression. The eukaryotic ncRNAs play a dual role shown to occur in a subset of inherited MEN1 tumors, in epigenetic regulation – some miRNAs can silence genes sporadic PNETs, sporadic pituitary tumors or sporadic that encode epigenetic factors such as DNMTs, and some parathyroid tumors at the promoter region of different lncRNAs can directly participate in chromatin-modifying genes (APC, CTNNB1, CDKN2A/p16, CDKN2B/p15, epigenetic control mechanisms such as in the chromatin HIC1, MEG3, MLH1, RASSF1A and RIZ1), with some also inhibitory complex polycomb group (PcG) repressive correlating with respective reduction of gene expression complex 2 (PRC2) (Fabbri et al. 2007, Kaneko et al. 2014, (Carling et al. 2003, Zhao et al. 2005, Lindberg et al. Chen et al. 2015, Mondal et al. 2015). 2008, Juhlin et al. 2010, Svedlund et al. 2012, Modali In endocrine cell-line models (MIN6 and βlox5 cells), et al. 2015). A genome-wide analysis of quantitative DNA microRNA-24 (miR24) has been shown to target menin, methylation in parathyroid tissue samples showed global and increased miR24 level was also shown to correlate DNA hypermethylation in menin-null parathyroid tumors with decreased menin level in the islets of Men1f/f;RIP-Cre from MEN1 patients, and increased transcript levels of mice and human parathyroid tumors (Luzi et al. 2012, DNMT1, predicting increased DNMT1 activity in menin- Vijayaraghavan et al. 2014, Grolmusz et al. 2017, Luzi et al. null parathyroid cells accounting for the hypermethylated 2017). This silencing of menin by a microRNA predicts DNA (Yuan et al. 2016). Another study showed altered DNA downstream effects on chromatin modifications because methylation profiles in normal and pathologic parathyroid menin acts as an epigenetic co-factor in chromatin- tissue with gene expression changes, and restored modifying protein complexes. Low levels of the lncRNA expression of hypermethylated genes upon treatment of MEG3 have been observed in mouse and human MEN1- primary cell cultures of parathyroid tumors with the DNA associated PNETs, human sporadic PNETs, and in human methylation inhibitor DAC (Starker et al. 2011). A few sporadic pituitary tumors (Zhao et al. 2005, Modali studies in endocrine/endocrine-related cell lines (BON1, et al. 2015). In MIN6 mouse insulinoma cells, Meg3 was H727, MIN6) have reported growth inhibition upon demonstrated to co-immunoprecipitate with Ezh2, which treatment with DNA methylation inhibitors 5AC or DAC is part of PRC2 for H3K27me3 establishment (Iyer et al. (Alexander et al. 2010, Modali et al. 2015). These studies 2017). How loss or gain of these ncRNA interactions suggest DNA hypermethylation as an oncogenic event affect endocrine tumor epigenetics should be further in MEN1-associated endocrine cell types, and therefore, investigated.

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R20 Endocrinology endocrine cells

Targeting the pathologic epigenome of Perspectives MEN1-associated endocrine cell types In this review, we aimed to survey the literature for Epigenome profiling for global epigenetic changes, such evidence of epigenetic regulation in ‘MEN1-associated as in DNA methylation or histone modifications between endocrine cell types’, which mainly included information normal and pathological samples, has demonstrated from the parathyroids, pituitary, pancreatic islets, the presence of epigenetic changes in tumors and other and lung neuroendocrine cells with the intention of disease states that could be exploited as targets for therapy highlighting the direct effects of menin as an epigenetic (Jones et al. 2016). Epigenetic therapies that are under co-factor or functionally related epigenetic effects. Whole- investigation to revert specific disease epigenomes to genome/exome sequencing efforts have identified somatic normal are designed to block the enzymatic activity of mutations in the MEN1 gene and/or genes associated with epigenetic regulators, disrupt interactions in chromatin- epigenetic control mechanisms in sporadic endocrine modifying protein complexes or target specific epigenetic tumors – non-functioning and functioning PNETs, factors for degradation (Ahuja et al. 2016). However, parathyroid adenoma and lung neuroendocrine tumors. designing of such therapeutics has to overcome several Future studies directed toward identifying the downstream issues and challenges such as cell-specific targeting due effects from these mutations will help to understand the to heterogeneity of the affected tissues, side effects, contribution of epigenetic changes in the pathogenesis of drug resistance and achieving constant, consistent and the specific endocrine tumors. Work conducted in cell lines long-lasting inhibition of the target (Dawson 2017). derived from pancreatic or lung neuroendocrine tumors Potential therapies in the form of various small molecule or their metastasis, and in vivo work in insulinomas of chemical inhibitors that target epigenetic enzymes (e.g., Men1f/f;RIP-Cre mice have established the role of menin as DNMTi, HDACi, KTMi, KDMi, and BETi) have been an epigenetic co-factor in the context of a few epigenetic developed. DNA hypomethylating agents (decitabine and modifications: H3K4me3, H3K9me3 and H4R3me2s and azacitidine) that target DNMT1 and DNMT3A, and a few DNA methylation. Given that DNA methylation and HDACi (panobinostat, vorinostat, and romidepsin) were multiple histone modifications are modulated by menin approved by the US Food and Drug Administration (FDA) raises the question whether epigenetic changes in MEN1- for the treatment of specific hematologic malignancies associated tumors would encompass multiple epigenetic (Ahuja et al. 2016). marks. Future studies directed toward determining the As discussed earlier, various HDACi, BETi and DNMTi changes in multiple histone modifications and/or DNA have been shown to inhibit the growth of endocrine/ methylation in tumors (epigenome signatures) compared endocrine-related cells lines, and intraperitoneal injection to corresponding normal cells could be valuable for of the BETi JQ1 has been shown to reduce cell proliferation understanding the tumorigenesis process and potential and increase apoptosis in insulinomas of the Men1f/f;RIP- treatment opportunities. The challenges for conducting Cre mouse model (Baradari et al. 2006, Adler et al. 2009, such studies for MEN1-associated tumors are the non- Alexander et al. 2010, Wong et al. 2014, Modali et al. availability of ‘corresponding’ specific normal cell types, 2015, Arvidsson et al. 2016, Lines et al. 2017). Another and the lack of relevant cell lines from human tumors. study showed that lack of a histone demethylase Rbp2 Perhaps generating mouse xenografts of human tumors in the Men1f/f;RIP-Cre mouse background could decrease could prove to be useful in establishing cell lines. A recent insulinoma development (Lin et al. 2011). Rbp2 loss study has shown that in the context of human PNETs could potentially restore H3K4me3 that occurred in the using a human hepatocyte growth factor (HGF) ligand insulinoma cells due to menin deficiency. Furthermore, facilitated the successful generation of xenografts in mice overexpression of RBP2 was detected in human PNETs because PNETs express high levels of the HGF receptor or their metastases, implicating a target for histone c-MET (Krampitz et al. 2016). Similar methods could demethylase inhibitor therapy (Maggi et al. 2016). These be used to establish cell lines from MEN1 and sporadic studies suggest the potential of epigenetic drug therapy tumors. Such tumor models and cell lines will facilitate of tumors arising in MEN1-associated endocrine cell a deeper understanding of epigenetic changes during the types irrespective of whether the epigenetic status of the tumorigenesis process to exploit epigenetic therapy as tumor cells is a direct consequence of menin loss or a a treatment option for the tumors of MEN1-associated consequence of the tumorigenesis process. endocrine cell types.

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R21 Endocrinology endocrine cells

Busygina V, Suphapeetiporn K, Marek LR, Stowers RS, Xu T & Bale AE Declaration of interest 2004 Hypermutability in a Drosophila model for multiple endocrine The authors declare that there is no conflict of interest that could be neoplasia type 1. Human Molecular Genetics 13 2399–2408. (https:// perceived as prejudicing the impartiality of this review. doi.org/10.1093/hmg/ddh271) Cao Y, Gao Z, Li L, Jiang X, Shan A, Cai J, Peng Y, Li Y, Jiang X, Huang X, et al. 2013 Whole exome sequencing of insulinoma reveals recurrent T372R mutations in YY1. Nature Communications 4 2810. Funding (https://doi.org/10.1038/ncomms3810) Funding support for this review was from the Intramural Research Carling T, Du Y, Fang W, Correa P & Huang S 2003 Intragenic allelic loss Program of the NIH, National Institute of Diabetes and Digestive and and promoter hypermethylation of the RIZ1 tumor suppressor gene Kidney Diseases. in parathyroid tumors and pheochromocytomas. Surgery 134 932–939. (https://doi.org/10.1016/S0039-6060(03)00422-7) Cech TR & Steitz JA 2014 The noncoding RNA revolution-trashing old References rules to forge new ones. Cell 157 77–94. (https://doi.org/10.1016/j. cell.2014.03.008) Adler JT, Hottinger DG, Kunnimalaiyaan M & Chen H 2009 Cedar H & Bergman Y 2009 Linking DNA methylation and histone Combination therapy with histone deacetylase inhibitors and modification: patterns and paradigms. Nature Reviews: Genetics 10 lithium chloride: a novel treatment for carcinoid tumors. Annals of 295–304. (https://doi.org/10.1038/nrg2540) Surgical Oncology 16 481–486. (https://doi.org/10.1245/s10434-008- Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, 0194-6) Emmert-Buck MR, Debelenko LV, Zhuang Z, Lubensky IA, Liotta LA, Agarwal SK 2014 Exploring the tumors of multiple endocrine neoplasia et al. 1997 Positional cloning of the gene for multiple endocrine type 1 in mouse models for basic and preclinical studies. neoplasia-type 1. Science 276 404–407. (https://doi.org/10.1126/ International Journal of Endocrine Oncology 1 153–161. (https://doi. science.276.5311.404) org/10.2217/ije.14.16) Chen Z, Liu S, Tian L, Wu M, Ai F, Tang W, Zhao L, Ding J, Zhang L & Agarwal SK 2017a The future: genetics advances in MEN1 therapeutic Tang A 2015 miR-124 and miR-506 inhibit colorectal cancer approaches and management strategies. Endocrine-Related Cancer 24 progression by targeting DNMT3B and DNMT1. Oncotarget 6 T119–T134. (https://doi.org/10.1530/ERC-17-0199) 38139–38150. (https://doi.org/10.18632/oncotarget.5709) Agarwal SK 2017b Update on exploring the tumors of multiple Cheng X 2014 Structural and functional coordination of DNA and endocrine neoplasia type 1 in mouse models for basic and preclinical histone methylation. Cold Spring Harbor Perspectives in Biology 6 studies. International Journal of Endocrine Oncology 4 113–116. a018747. (https://doi.org/10.1101/cshperspect.a018747) (https://doi.org/10.2217/ije-2017-0013) Chi P, Allis CD & Wang GG 2010 Covalent histone modifications – Agarwal SK & Jothi R 2012 Genome-wide characterization of menin- miswritten, misinterpreted and mis-erased in human cancers. dependent H3K4me3 reveals a specific role for menin in the Nature Reviews Cancer 10 457–469. (https://doi.org/10.1038/ regulation of genes implicated in MEN1-like tumors. PLoS ONE 7 nrc2876) e37952. (https://doi.org/10.1371/journal.pone.0037952) Chou WC, Lin PH, Yeh YC, Shyr YM, Fang WL, Wang SE, Liu CY, Ahuja N, Sharma AR & Baylin SB 2016 Epigenetic therapeutics: a new Chang PM, Chen MH, Hung YP, et al. 2016 Genes involved in weapon in the war against cancer. Annual Review of Medicine 67 angiogenesis and mTOR pathways are frequently mutated in Asian 73–89. (https://doi.org/10.1146/annurev-med-111314-035900) patients with pancreatic neuroendocrine tumors. International Journal Alexander VM, Roy M, Steffens KA, Kunnimalaiyaan M & Chen H 2010 of Biological Sciences 12 1523–1532. (https://doi.org/10.7150/ Azacytidine induces cell cycle arrest and suppression of ijbs.16233) neuroendocrine markers in carcinoids. International Journal of Clinical Clapier CR & Cairns BR 2009 The biology of chromatin remodeling and Experimental Medicine 3 95–102. complexes. Annual Review of Biochemistry 78 273–304. (https://doi. Allis CD & Jenuwein T 2016 The molecular hallmarks of epigenetic org/10.1146/annurev.biochem.77.062706.153223) control. Nature Reviews: Genetics 17 487–500. (https://doi. Cromer MK, Starker LF, Choi M, Udelsman R, Nelson-Williams C, org/10.1038/nrg.2016.59) Lifton RP & Carling T 2012 Identification of somatic mutations in Allis CD, Caparros M-L, Jenuwein T & Reinberg D 2015 Epigenetics. Cold parathyroid tumors using whole-exome sequencing. Journal of Spring Harbor, New York: CSH Press, Cold Spring Harbor Laboratory Clinical Endocrinology and Metabolism 97 E1774–E1781. (https://doi. Press. org/10.1210/jc.2012-1743) Amorim JP, Santos G, Vinagre J & Soares P 2016 The role of ATRX in the Dawson MA 2017 The cancer epigenome: concepts, challenges, and alternative lengthening of telomeres (ALT) phenotype. Genes 766. therapeutic opportunities. Science 355 1147–1152. (https://doi. (https://doi.org/10.3390/genes7090066) org/10.1126/science.aam7304) Arvidsson Y, Johanson V, Pfragner R, Wangberg B & Nilsson O 2016 Debelenko LV, Brambilla E, Agarwal SK, Swalwell JI, Kester MB, Cytotoxic effects of valproic acid on neuroendocrine tumour cells. Lubensky IA, Zhuang Z, Guru SC, Manickam P, Olufemi SE, et al. Neuroendocrinology 103 578–591. (https://doi.org/10.1159/000441849) 1997 Identification of MEN1 gene mutations in sporadic carcinoid Baradari V, Huether A, Hopfner M, Schuppan D & Scherubl H 2006 tumors of the lung. Human Molecular Genetics 6 2285–2290. (https:// Antiproliferative and proapoptotic effects of histone deacetylase doi.org/10.1093/hmg/6.13.2285) inhibitors on gastrointestinal neuroendocrine tumor cells. Endocrine- Dreijerink KMA, Timmers HTM & Brown M 2017 Twenty years of Related Cancer 13 1237–1250. (https://doi.org/10.1677/erc.1.01249) menin: emerging opportunities for restoration of transcriptional Barlow DP & Bartolomei MS 2014 Genomic imprinting in mammals. regulation in MEN1. Endocrine-Related Cancer 24 T135–T145. (https:// Cold Spring Harbor Perspectives in Biology 6. doi.org/10.1530/ERC-17-0281) Bi WL, Horowitz P, Greenwald NF, Abedalthagafi M, Agarwalla PK, Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, Liu S, Gibson WJ, Mei Y, Schumacher SE, Ben-David U, Chevalier A, et al. Alder H, Costinean S, Fernandez-Cymering C, et al. 2007 2017 Landscape of genomic alterations in pituitary adenomas. MicroRNA-29 family reverts aberrant methylation in lung cancer by Clinical Cancer Research 23 1841–1851. (https://doi. targeting DNA methyltransferases 3A and 3B. PNAS 104 org/10.1158/1078-0432.CCR-16-0790) 15805–15810. (https://doi.org/10.1073/pnas.0707628104)

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R22 Endocrinology endocrine cells

Feng Z, Ma J & Hua X 2017a Epigenetic regulation by the menin Karnik SK, Hughes CM, Gu X, Rozenblatt-Rosen O, McLean GW, pathway. Endocrine-Related Cancer 24 T147–T159. (https://doi. Xiong Y, Meyerson M & Kim SK 2005 Menin regulates pancreatic org/10.1530/ERC-17-0298) islet growth by promoting histone methylation and expression of Feng Z, Wang L, Sun Y, Jiang Z, Domsic J, An C, Xing B, Tian J, Liu X, genes encoding p27Kip1 and p18INK4c. PNAS 102 14659–14664. Metz DC, et al. 2017b Menin and Daxx interact to suppress (https://doi.org/10.1073/pnas.0503484102) neuroendocrine tumors through epigenetic control of the membrane Kim JY, Brosnan-Cashman JA, An S, Kim SJ, Song KB, Kim MS, Kim MJ, metallo-endopeptidase. Cancer Research 77 401–411. (https://doi. Hwang DW, Meeker AK, Yu E, et al. 2017 Alternative lengthening of org/10.1158/0008-5472.CAN-16-1567) telomeres in primary pancreatic neuroendocrine tumors is associated Flavahan WA, Gaskell E & Bernstein BE 2017 Epigenetic plasticity and with aggressive clinical behavior and poor survival. Clinical Cancer the hallmarks of cancer. Science 357 eaal2380. (https://doi. Research 23 1598–1606. (https://doi.org/10.1158/1078-0432.CCR-16- org/10.1126/science.aal2380) 1147) Greenblatt DY, Vaccaro AM, Jaskula-Sztul R, Ning L, Haymart M, Kopp F & Mendell JT 2018 Functional classification and experimental Kunnimalaiyaan M & Chen H 2007 Valproic acid activates notch-1 dissection of long noncoding RNAs. Cell 172 393–407. (https://doi. signaling and regulates the neuroendocrine phenotype in carcinoid org/10.1016/j.cell.2018.01.011) cancer cells. Oncologist 12 942–951. (https://doi.org/10.1634/ Koschmann C, Nunez FJ, Mendez F, Brosnan-Cashman JA, Meeker AK, theoncologist.12-8-942) Lowenstein PR & Castro MG 2017 Mutated chromatin regulatory Grolmusz VK, Borka K, Kovesdi A, Nemeth K, Balogh K, Dekany C, factors as tumor drivers in cancer. Cancer Research 77 227–233. Kiss A, Szentpeteri A, Sarman B, Somogyi A, et al. 2017 MEN1 (https://doi.org/10.1158/0008-5472.CAN-16-2301) mutations and potentially MEN1-targeting miRNAs are responsible Krampitz GW, George BM, Willingham SB, Volkmer JP, Weiskopf K, for menin deficiency in sporadic and MEN1 syndrome-associated Jahchan N, Newman AM, Sahoo D, Zemek AJ, Yanovsky RL, et al. primary hyperparathyroidism. Virchows Archiv 471 401–411. (https:// 2016 Identification of tumorigenic cells and therapeutic targets in doi.org/10.1007/s00428-017-2158-3) pancreatic neuroendocrine tumors. PNAS 113 4464–4469. (https:// Gurung B, Feng Z, Iwamoto DV, Thiel A, Jin G, Fan CM, Ng JM, doi.org/10.1073/pnas.1600007113) Curran T & Hua X 2013 Menin epigenetically represses Hedgehog Lemmens I, Van de Ven WJ, Kas K, Zhang CX, Giraud S, Wautot V, signaling in MEN1 tumor syndrome. Cancer Research 73 2650–2658. Buisson N, De Witte K, Salandre J, Lenoir G, et al. 1997 (https://doi.org/10.1158/0008-5472.CAN-12-3158) Identification of the multiple endocrine neoplasia type 1 (MEN1) Hendy GN, Kaji H & Canaff L 2009 Cellular functions of menin. In gene. The European Consortium on MEN1. Human Molecular Genetics SuperMEN1 Pituitary, Parathyroid and Pancreas. pp 37–50. Eds. K 6 1177–1183. (https://doi.org/10.1093/hmg/6.7.1177) Balogh & A Patocs. New York: Springer, New York, NY (https://doi. Li E & Zhang Y 2014 DNA methylation in mammals. Cold Spring Harbor org/10.1007/978-1-4419-1664-8) Perspectives in Biology 6 a019133. (https://doi.org/10.1101/ Huang J, Gurung B, Wan B, Matkar S, Veniaminova NA, Wan K, cshperspect.a019133) Merchant JL, Hua X & Lei M 2012 The same pocket in menin binds Lichtenauer UD, Di Dalmazi G, Slater EP, Wieland T, Kuebart A, both MLL and JUND but has opposite effects on transcription. Nature Schmittfull A, Schwarzmayr T, Diener S, Wiese D, Thasler WE, et al. 482 542.e451–546.e451. (https://doi.org/10.1038/nature10806) 2015 Frequency and clinical correlates of somatic Ying Yang 1 Huang H, Sabari BR, Garcia BA, Allis CD & Zhao Y 2014 SnapShot: mutations in sporadic insulinomas. Journal of Clinical Endocrinology histone modifications. Cell 159 458.e451. (https://doi.org/10.1016/j. and Metabolism 100 E776–E782. (https://doi.org/10.1210/jc.2015- cell.2014.09.037) 1100) Hughes CM, Rozenblatt-Rosen O, Milne TA, Copeland TD, Levine SS, Lin W, Cao J, Liu J, Beshiri ML, Fujiwara Y, Francis J, Cherniack AD, Lee JC, Hayes DN, Shanmugam KS, Bhattacharjee A, Biondi CA, et al. Geisen C, Blair LP, Zou MR, et al. 2011 Loss of the retinoblastoma 2004 Menin associates with a trithorax family histone binding protein 2 (RBP2) histone demethylase suppresses methyltransferase complex and with the hoxc8 locus. Molecular Cell tumorigenesis in mice lacking Rb1 or Men1. PNAS 108 13 587.e451–597.e451. (https://doi.org/10.1016/S1097- 13379–13386. (https://doi.org/10.1073/pnas.1110104108) 2765(04)00081-4) Lin W, Watanabe H, Peng S, Francis JM, Kaplan N, Pedamallu CS, Iyer S, Modali SD & Agarwal SK 2017 The long non-coding RNA MEG3 Ramachandran A, Agoston A, Bass AJ & Meyerson M 2015 Dynamic is an epigenetic determinant of oncogenic signaling in functional epigenetic regulation by menin during pancreatic islet tumor pancreatic neuroendocrine tumor cells. Molecular and Cellular Biology formation. Molecular Cancer Research 13 689–698. (https://doi. 37 e00278-17. (https://doi.org/10.1128/MCB.00278-17) org/10.1158/1541-7786.MCR-14-0457) Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, Schulick RD, Lin W, Francis JM, Li H, Gao X, Pedamallu CS, Ernst P & Meyerson M Tang LH, Wolfgang CL, Choti MA, et al. 2011 DAXX/ATRX, MEN1, 2016 Kmt2a cooperates with menin to suppress tumorigenesis in and mTOR pathway genes are frequently altered in pancreatic mouse pancreatic islets. Cancer Biology and Therapy 17 1274–1281. neuroendocrine tumors. Science 331 1199.e451–1203.e451. (https:// (https://doi.org/10.1080/15384047.2016.1250986) doi.org/10.1126/science.1200609) Lindberg D, Akerstrom G & Westin G 2008 Evaluation of CDKN2C/p18, Jones PA, Issa JP & Baylin S 2016 Targeting the cancer epigenome for CDKN1B/p27 and CDKN2B/p15 mRNA expression, and CpG therapy. Nature Reviews: Genetics 17 630–641. (https://doi. methylation status in sporadic and MEN1-associated pancreatic org/10.1038/nrg.2016.93) endocrine tumours. Clinical Endocrinology 68 271–277. (https://doi. Juhlin CC, Kiss NB, Villablanca A, Haglund F, Nordenstrom J, Hoog A & org/10.1111/j.1365-2265.2007.03034.x) Larsson C 2010 Frequent promoter hypermethylation of the APC Lines KE, Stevenson M, Filippakopoulos P, Muller S, Lockstone HE, and RASSF1A tumour suppressors in parathyroid tumours. PLoS ONE Wright B, Grozinsky-Glasberg S, Grossman AB, Knapp S, Buck D, et 5 e9472. (https://doi.org/10.1371/journal.pone.0009472) al. 2017 Epigenetic pathway inhibitors represent potential drugs for Kaku M, Nishiyama T, Yagawa K & Abe M 1980 Establishment of a treating pancreatic and bronchial neuroendocrine tumors. carcinoembryonic antigen-producing cell line from human Oncogenesis 6 e332. (https://doi.org/10.1038/oncsis.2017.30) pancreatic carcinoma. Gan 71 596–601. Luzi E, Marini F, Giusti F, Galli G, Cavalli L & Brandi ML 2012 The Kaneko S, Bonasio R, Saldana-Meyer R, Yoshida T, Son J, Nishino K, negative feedback-loop between the oncomir Mir-24-1 and menin Umezawa A & Reinberg D 2014 Interactions between JARID2 and modulates the Men1 tumorigenesis by mimicking the ‘Knudson’s noncoding RNAs regulate PRC2 recruitment to chromatin. Molecular second hit’. PLoS ONE 7 e39767. (https://doi.org/10.1371/journal. Cell 53 290–300. (https://doi.org/10.1016/j.molcel.2013.11.012) pone.0039767)

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R23 Endocrinology endocrine cells

Luzi E, Ciuffi S, Marini F, Mavilia C, Galli G & Brandi ML 2017 Analysis Poncin J, Stevenaert A & Beckers A 1999 Somatic MEN1 gene mutation of differentially expressed microRNAs in MEN1 parathyroid does not contribute significantly to sporadic pituitary tumorigenesis. adenomas. American Journal of Translational Research 9 1743–1753. European Journal of Endocrinology 140 573–576. (https://doi. Maggi EC, Trillo-Tinoco J, Struckhoff AP, Vijayaraghavan J, Del Valle L & org/10.1530/eje.0.1400573) Crabtree JS 2016 Retinoblastoma-binding protein 2 (RBP2) is Romano R, Soong CP, Rose M, Costa-Guda J, Bellizzi J & Arnold A 2017 frequently expressed in neuroendocrine tumors and promotes the EZH2 copy number and mutational analyses in sporadic parathyroid neoplastic phenotype. Oncogenesis 5 e257. (https://doi.org/10.1038/ adenomas. Endocrine 55 985–988. (https://doi.org/10.1007/s12020- oncsis.2016.58) 016-1142-z) Marini F, Giusti F, Tonelli F & Brandi ML 2017 Management impact: Rowley MJ & Corces VG 2016 The three-dimensional genome: principles effects on quality of life and prognosis in MEN1. Endocrine-Related and roles of long-distance interactions. Current Opinion in Cell Biology Cancer 24 T227–T242. (https://doi.org/10.1530/ERC-17-0203) 40 8–14. (https://doi.org/10.1016/j.ceb.2016.01.009) Marinoni I, Wiederkeher A, Wiedmer T, Pantasis S, Di Domenico A, Sato T, Issa JJ & Kropf P 2017 DNA hypomethylating drugs in cancer Frank R, Vassella E, Schmitt A & Perren A 2017 Hypo-methylation therapy. Cold Spring Harbor Perspectives in Medicine 7 a026948. mediates chromosomal instability in pancreatic NET. Endocrine- (https://doi.org/10.1101/cshperspect.a026948) Related Cancer 24 137–146. (https://doi.org/10.1530/ERC-16-0554) Scacheri PC, Davis S, Odom DT, Crawford GE, Perkins S, Halawi MJ, Marmorstein R & Zhou MM 2014 Writers and readers of histone Agarwal SK, Marx SJ, Spiegel AM, Meltzer PS, et al. 2006 Genome- acetylation: structure, mechanism, and inhibition. Cold Spring Harbor wide analysis of menin binding provides insights into MEN1 Perspectives in Biology 6 a018762. (https://doi.org/10.1101/ tumorigenesis. PLoS Genetics 2 e51. (https://doi.org/10.1371/journal. cshperspect.a018762) pgen.0020051) Marques P & Korbonits M 2017 Genetic aspects of pituitary adenomas. Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, Lawlor RT, Endocrinology Metabolism Clinics of North America 46 335–374. Johns AL, Miller DK, Mafficini A, et al. 2017 Whole-genome (https://doi.org/10.1016/j.ecl.2017.01.004) landscape of pancreatic neuroendocrine tumours. Nature 543 65–71. Martinelli DC & Fan CM 2007 The role of Gas1 in embryonic (https://doi.org/10.1038/nature21063) development and its implications for human disease. Cell Cycle 6 Schussheim DH, Skarulis MC, Agarwal SK, Simonds WF, Burns AL, 2650–2655. (https://doi.org/10.4161/cc.6.21.4877) Spiegel AM & Marx SJ 2001 Multiple endocrine neoplasia type 1: Merglen A, Theander S, Rubi B, Chaffard G, Wollheim CB & new clinical and basic findings. Trends in Endocrinology and Maechler P 2004 Glucose sensitivity and metabolism-secretion Metabolism 12 173–178. (https://doi.org/10.1016/S1043- coupling studied during two-year continuous culture in INS-1E 2760(00)00372-6) insulinoma cells. Endocrinology 145 667–678. (https://doi. Seto E & Yoshida M 2014 Erasers of histone acetylation: the histone org/10.1210/en.2003-1099) deacetylase enzymes. Cold Spring Harbor Perspectives in Biology 6 Miyazaki J, Araki K, Yamato E, Ikegami H, Asano T, Shibasaki Y, Oka Y & a018713. (https://doi.org/10.1101/cshperspect.a018713) Yamamura K 1990 Establishment of a pancreatic beta cell line that Shilatifard A 2012 The COMPASS family of histone H3K4 methylases: retains glucose-inducible insulin secretion: special reference to mechanisms of regulation in development and disease pathogenesis. expression of glucose transporter isoforms. Endocrinology 127 Annual Review of Biochemistry 81 65–95. (https://doi.org/10.1146/ 126–132. (https://doi.org/10.1210/endo-127-1-126) annurev-biochem-051710-134100) Modali SD, Parekh VI, Kebebew E & Agarwal SK 2015 Epigenetic Simbolo M, Mafficini A, Sikora KO, Fassan M, Barbi S, Corbo V, regulation of the lncRNA MEG3 and its target c-MET in pancreatic Mastracci L, Rusev B, Grillo F, Vicentini C, et al. 2017 Lung neuroendocrine tumors. Molecular Endocrinology 29 224–237. (https:// neuroendocrine tumours: deep sequencing of the four World Health doi.org/10.1210/me.2014-1304) Organization histotypes reveals chromatin-remodelling genes as Mondal T, Subhash S, Vaid R, Enroth S, Uday S, Reinius B, Mitra S, major players and a prognostic role for TERT, RB1, MEN1 and Mohammed A, James AR, Hoberg E, et al. 2015 MEG3 long KMT2D. Journal of Pathology 241 488–500. (https://doi.org/10.1002/ noncoding RNA regulates the TGF-beta pathway genes through path.4853) formation of RNA-DNA triplex structures. Nature Communications 6 Singhi AD, Liu TC, Roncaioli JL, Cao D, Zeh HJ, Zureikat AH, Tsung A, 7743. (https://doi.org/10.1038/ncomms8743) Marsh JW, Lee KK, Hogg ME, et al. 2017 Alternative lengthening of Muntean AG & Hess JL 2012 The pathogenesis of mixed-lineage telomeres and loss of DAXX/ATRX expression predicts metastatic leukemia. Annual Review of Pathology 7 283–301. (https://doi. disease and poor survival in patients with pancreatic neuroendocrine org/10.1146/annurev-pathol-011811-132434) tumors. Clinical Cancer Research 23 600–609. (https://doi. Murai MJ, Chruszcz M, Reddy G, Grembecka J & Cierpicki T 2011 org/10.1158/1078-0432.CCR-16-1113) Crystal structure of menin reveals binding site for mixed lineage Starker LF, Svedlund J, Udelsman R, Dralle H, Akerstrom G, Westin G, leukemia (MLL) protein. Journal of Biological Chemistry 286 Lifton RP, Bjorklund P & Carling T 2011 The DNA methylome of 31742–31748. (https://doi.org/10.1074/jbc.M111.258186) benign and malignant parathyroid tumors. Genes Newey PJ, Nesbit MA, Rimmer AJ, Attar M, Head RT, Christie PT, Cancer 50 735–745. (https://doi.org/10.1002/gcc.20895) Gorvin CM, Stechman M, Gregory L, Mihai R, et al. 2012 Whole- Stopa N, Krebs JE & Shechter D 2015 The PRMT5 arginine exome sequencing studies of nonhereditary (sporadic) parathyroid methyltransferase: many roles in development, cancer and beyond. adenomas. Journal of Clinical Endocrinology and Metabolism 97 Cellular and Molecular Life Sciences 72 2041–2059. (https://doi. E1995–E2005. (https://doi.org/10.1210/jc.2012-2303) org/10.1007/s00018-015-1847-9) Newey PJ, Nesbit MA, Rimmer AJ, Head RA, Gorvin CM, Attar M, Svedlund J, Koskinen Edblom S, Marquez VE, Akerstrom G, Bjorklund P & Gregory L, Wass JA, Buck D, Karavitaki N, et al. 2013 Whole-exome Westin G 2012 Hypermethylated in cancer 1 (HIC1), a tumor sequencing studies of nonfunctioning pituitary adenomas. Journal of suppressor gene epigenetically deregulated in hyperparathyroid tumors Clinical Endocrinology and Metabolism 98 E796–E800. (https://doi. by histone H3 lysine modification. Journal of Clinical Endocrinology and org/10.1210/jc.2012-4028) Metabolism 97 E1307–E1315. (https://doi.org/10.1210/jc.2011-3136) Olins DE & Olins AL 2003 Chromatin history: our view from the bridge. Szenker E, Ray-Gallet D & Almouzni G 2011 The double face of the Nature Reviews: Molecular Cell Biology 4 809–814. (https://doi. histone variant H3.3. Cell Research 21 421–434. (https://doi. org/10.1038/nrm1225) org/10.1038/cr.2011.14) Perez-Salvia M & Esteller M 2017 Bromodomain inhibitors and cancer Thakker RV, Newey PJ, Walls GV, Bilezikian J, Dralle H, Ebeling PR, therapy: from structures to applications. Epigenetics 12 323–339. Melmed S, Sakurai A, Tonelli F, Brandi ML, et al. 2012 Clinical (https://doi.org/10.1080/15592294.2016.1265710) practice guidelines for multiple endocrine neoplasia type 1 (MEN1).

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access Journal of Molecular S Iyer and S K Agarwal Epigenetic regulation in 61:1 R24 Endocrinology endocrine cells

Journal of Clinical Endocrinology and Metabolism 97 2990–3011. Yang YJ, Song TY, Park J, Lee J, Lim J, Jang H, Kim YN, Yang JH, Song Y, (https://doi.org/10.1210/jc.2012-1230) Choi A, et al. 2013 Menin mediates epigenetic regulation via histone Townsend CM Jr, Ishizuka J & Thompson JC 1993 Studies of growth H3 lysine 9 methylation. Cell Death and Disease 4 e583. (https://doi. regulation in a neuroendocrine cell line. Acta Oncologica 32 125–130. org/10.1038/cddis.2013.98) (https://doi.org/10.3109/02841869309083900) Yokoyama A, Wang Z, Wysocka J, Sanyal M, Aufiero DJ, Kitabayashi I, van Nuland R, Smits AH, Pallaki P, Jansen PW, Vermeulen M & Herr W & Cleary ML 2004 Leukemia proto-oncoprotein MLL forms a Timmers HT 2013 Quantitative dissection and stoichiometry SET1-like histone methyltransferase complex with menin to regulate determination of the human SET1/MLL histone methyltransferase Hox gene expression. Molecular and Cellular Biology 24 5639–5649. complexes. Molecular and Cellular Biology 33 2067–2077. (https://doi. (https://doi.org/10.1128/MCB.24.13.5639-5649.2004) org/10.1128/MCB.01742-12) Yokoyama A, Somervaille TC, Smith KS, Rozenblatt-Rosen O, VandenBussche CJ, Allison DB, Graham MK, Charu V, Lennon AM, Meyerson M & Cleary ML 2005 The menin tumor suppressor protein Wolfgang CL, Hruban RH & Heaphy CM 2017 Alternative is an essential oncogenic cofactor for MLL-associated lengthening of telomeres and ATRX/DAXX loss can be reliably leukemogenesis. Cell 123 207–218. (https://doi.org/10.1016/j. detected in FNAs of pancreatic neuroendocrine tumors. Cancer cell.2005.09.025) Cytopathology 125 544–551. (https://doi.org/10.1002/cncy.21857) Yuan Z, Sanchez Claros C, Suzuki M, Maggi EC, Kaner JD, Kinstlinger N, Vijayaraghavan J, Maggi EC & Crabtree JS 2014 miR-24 regulates menin Gorecka J, Quinn TJ, Geha R, Corn A, et al. 2016 Loss of MEN1 in the endocrine pancreas. American Journal of Physiology: activates DNMT1 implicating DNA hypermethylation as a driver of Endocrinology and Metabolism 307 E84–E92. (https://doi.org/10.1152/ MEN1 tumorigenesis. Oncotarget 7 12633–12650. (https://doi. ajpendo.00542.2013) org/10.18632/oncotarget.7279) Voigt P, Tee WW & Reinberg D 2013 A double take on bivalent Zhao J, Dahle D, Zhou Y, Zhang X & Klibanski A 2005 promoters. Genes and Development 27 1318–1338. (https://doi. Hypermethylation of the promoter region is associated with the loss org/10.1101/gad.219626.113) of MEG3 gene expression in human pituitary tumors. Journal of Wang H, Bender A, Wang P, Karakose E, Inabnet WB, Libutti SK, Clinical Endocrinology and Metabolism 90 2179–2186. (https://doi. Arnold A, Lambertini L, Stang M, Chen H, et al. 2017 Insights into org/10.1210/jc.2004-1848) beta cell regeneration for diabetes via integration of molecular Zhao J, Zhang X, Zhou Y, Ansell PJ & Klibanski A 2006 Cyclic AMP landscapes in human insulinomas. Nature Communications 8 767. stimulates MEG3 gene expression in cells through a cAMP-response (https://doi.org/10.1038/s41467-017-00992-9) element (CRE) in the MEG3 proximal promoter region. International Wong C, Laddha SV, Tang L, Vosburgh E, Levine AJ, Normant E, Journal of Biochemistry and Cell Biology 38 1808–1820. (https://doi. Sandy P, Harris CR, Chan CS & Xu EY 2014 The bromodomain and org/10.1016/j.biocel.2006.05.004) extra-terminal inhibitor CPI203 enhances the antiproliferative effects Zhuang Z, Vortmeyer AO, Pack S, Huang S, Pham TA, Wang C, Park WS, of rapamycin on human neuroendocrine tumors. Cell Death and Agarwal SK, Debelenko LV, Kester M, et al. 1997 Somatic mutations Disease 5 e1450. (https://doi.org/10.1038/cddis.2014.396) of the MEN1 tumor suppressor gene in sporadic gastrinomas and Wu T & Hua X 2011 Menin represses tumorigenesis via repressing cell insulinomas. Cancer Research 57 4682–4686. proliferation. American Journal of Cancer Research 1 726–739. Zoghbi HY & Beaudet AL 2016 Epigenetics and human disease. Cold Yang Y & Bedford MT 2013 Protein arginine methyltransferases and cancer. Spring Harbor Perspectives in Biology 8 a019497. (https://doi. Nature Reviews Cancer 13 37–50. (https://doi.org/10.1038/nrc3409) org/10.1101/cshperspect.a01949)

Received 18 February 2018 Accepted 3 April 2018 Accepted Preprint published online 3 April 2018

http://jme.endocrinology-journals.org © 2018 Society for Endocrinology https://doi.org/10.1530/JME-18-0050 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 10/03/2021 12:22:59PM via free access